Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
J Neurosci Res ; 102(6): e25360, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38847288

RESUMEN

Childhood obesity increases the risk of health and cognitive disorders in adulthood. Consuming high-fat diets (HFD) during critical neurodevelopmental periods, like childhood, impairs cognition and memory in humans and animals, affecting the function and connectivity of brain structures related to emotional memory. However, the underlying mechanisms of such phenomena need to be better understood. This study aimed to investigate the neurochemical profile of the amygdala and hippocampus, brain structures involved in emotional memory, during the acquisition of conditioned odor aversion in male rats that consumed a HFD from weaning to adulthood. The rats gained weight, experienced metabolic changes, and reduced insulin sensitivity and glucose tolerance. Rats showed enhanced odor aversion memory, contrary to the expected cognitive impairments. This memory enhancement was accompanied by increased noradrenergic and glutamatergic neurotransmission in the amygdala and hippocampus. Importantly, this upregulation was specific to stimuli exposure, as basal neurotransmitter levels remained unaltered by the HFD. Our results suggest that HFD modifies cognitive function by altering neurochemical signaling, in this case, upregulating neurotransmitter levels rendering a stronger memory trace, demonstrating that metabolic dysfunctions do not only trigger exclusively detrimental plasticity processes but also render enhanced plastic effects depending on the type of information.


Asunto(s)
Amígdala del Cerebelo , Dieta Alta en Grasa , Ácido Glutámico , Hipocampo , Transmisión Sináptica , Animales , Masculino , Dieta Alta en Grasa/efectos adversos , Hipocampo/metabolismo , Amígdala del Cerebelo/metabolismo , Transmisión Sináptica/fisiología , Ratas , Ácido Glutámico/metabolismo , Norepinefrina/metabolismo , Ratas Wistar , Cognición/fisiología , Reacción de Prevención/fisiología
2.
Cell Mol Life Sci ; 77(18): 3469-3502, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32006052

RESUMEN

Throughout the animal kingdom sucrose is one of the most palatable and preferred tastants. From an evolutionary perspective, this is not surprising as it is a primary source of energy. However, its overconsumption can result in obesity and an associated cornucopia of maladies, including type 2 diabetes and cardiovascular disease. Here we describe three physiological levels of processing sucrose that are involved in the decision to ingest it: the tongue, gut, and brain. The first section describes the peripheral cellular and molecular mechanisms of sweet taste identification that project to higher brain centers. We argue that stimulation of the tongue with sucrose triggers the formation of three distinct pathways that convey sensory attributes about its quality, palatability, and intensity that results in a perception of sweet taste. We also discuss the coding of sucrose throughout the gustatory pathway. The second section reviews how sucrose, and other palatable foods, interact with the gut-brain axis either through the hepatoportal system and/or vagal pathways in a manner that encodes both the rewarding and of nutritional value of foods. The third section reviews the homeostatic, hedonic, and aversive brain circuits involved in the control of food intake. Finally, we discuss evidence that overconsumption of sugars (or high fat diets) blunts taste perception, the post-ingestive nutritional reward value, and the circuits that control feeding in a manner that can lead to the development of obesity.


Asunto(s)
Obesidad/patología , Azúcares/metabolismo , Gusto/fisiología , Animales , Encéfalo/metabolismo , Humanos , Leptina/metabolismo , Neuronas/metabolismo , Valor Nutritivo , Obesidad/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
3.
J Neurosci ; 36(50): 12511-12529, 2016 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-27974611

RESUMEN

Although the release of mesoaccumbal dopamine is certainly involved in rewarding responses, recent studies point to the importance of the interaction between it and glutamate. One important component of this network is the anterior nucleus accumbens shell (aNAcSh), which sends GABAergic projections into the lateral hypothalamus (LH) and receives extensive glutamatergic inputs from, among others, the medial prefrontal cortex (mPFC). The effects of glutamatergic activation of aNAcSh on the ingestion of rewarding stimuli as well as its effect in the LH and mPFC are not well understood. Therefore, we studied behaving mice that express a light-gated channel (ChR2) in glutamatergic fibers in their aNAcSh while recording from neurons in the aNAcSh, or mPFC or LH. In Thy1-ChR2, but not wild-type, mice activation of aNAcSh fibers transiently stopped the mice licking for sucrose or an empty sipper. Stimulation of aNAcSh fibers both activated and inhibited single-unit responses aNAcSh, mPFC, and LH, in a manner that maintains firing rate homeostasis. One population of licking-inhibited pMSNs in the aNAcSh was also activated by optical stimulation, suggesting their relevance in the cessation of feeding. A rewarding aspect of stimulation of glutamatergic inputs was found when the Thy1-ChR2 mice learned to nose-poke to self-stimulate these inputs, indicating that bulky stimulation of these fibers are rewarding in the sense of wanting. Stimulation of excitatory afferents evoked both monosynaptic and polysynaptic responses distributed in the three recorded areas. In summary, we found that activation of glutamatergic aNAcSh fibers is both rewarding and transiently inhibits feeding. SIGNIFICANCE STATEMENT: We have established that the activation of glutamatergic fibers in the anterior nucleus accumbens shell (aNAcSh) transiently stops feeding and yet, because mice self-stimulate, is rewarding in the sense of wanting. Moreover, we have characterized single-unit responses of distributed components of a hedonic network (comprising the aNAcSh, medial prefrontal cortex, and lateral hypothalamus) recruited by activation of glutamatergic aNAcSh afferents that are involved in encoding a positive valence signal important for the wanting of a reward and that transiently stops ongoing consummatory actions, such as licking.


Asunto(s)
Conducta Alimentaria/fisiología , Glutamatos/fisiología , Área Hipotalámica Lateral/fisiología , Fibras Nerviosas/fisiología , Núcleo Accumbens/citología , Núcleo Accumbens/fisiología , Corteza Prefrontal/fisiología , Recompensa , Animales , Channelrhodopsins , Femenino , Masculino , Ratones , Neuronas Aferentes/fisiología , Optogenética , Técnicas de Placa-Clamp , Autoestimulación , Sinapsis/fisiología
4.
Appetite ; 100: 152-61, 2016 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-26867698

RESUMEN

Obesity is a public health problem caused by excessive consumption of high caloric diets and/or lack of physical activity. Although treatments for obesity include low caloric diets and exercise programs, these activities frequently are supplemented with appetite suppressants. For the short-term treatment of weight loss, diethylpropion (DEP) is a commonly used appetite suppressant. However, little is known with regard to how to improve its weight loss efficacy. We therefore evaluated, in rats, two administration protocols where the animals received daily injections of DEP. First, when these nocturnal animals were normally active (at night) and when they were normally inactive (daytime), and second, with or without high fat dietary restriction (HFDR). We observed that DEP induced a greater weight-loss administered when the animals were in their active phase than in their inactive phase. Moreover, DEP's administration during the inactive phase (and to a lesser degree in the active phase) promotes the consumption of food during normal sleeping time. In addition, we found that DEP-induced weight loss under ad libitum access to a HF diet, but its efficacy significantly improved under conditions of HFDR. In summary, the efficacy of DEP, and presumably other like appetite suppressants, is enhanced by carefully controlling the time it is administered and under dietary restriction of HF diets.


Asunto(s)
Depresores del Apetito/uso terapéutico , Regulación del Apetito/efectos de los fármacos , Dieta con Restricción de Grasas , Dieta Reductora , Dietilpropión/uso terapéutico , Sobrepeso/tratamiento farmacológico , Pérdida de Peso/efectos de los fármacos , Animales , Depresores del Apetito/administración & dosificación , Depresores del Apetito/efectos adversos , Depresores del Apetito/farmacocinética , Biotransformación , Ritmo Circadiano/efectos de los fármacos , Terapia Combinada/efectos adversos , Dieta Alta en Grasa/efectos adversos , Dietilpropión/administración & dosificación , Dietilpropión/efectos adversos , Dietilpropión/análogos & derivados , Dietilpropión/sangre , Dietilpropión/farmacocinética , Esquema de Medicación , Ingestión de Energía/efectos de los fármacos , Semivida , Inyecciones Intraperitoneales , Masculino , Sobrepeso/sangre , Sobrepeso/dietoterapia , Sobrepeso/etiología , Fenilpropanolamina/análogos & derivados , Fenilpropanolamina/sangre , Ratas Sprague-Dawley
5.
J Neurophysiol ; 114(1): 585-607, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25972577

RESUMEN

Obesity is a worldwide health problem that has reached epidemic proportions. To ameliorate this problem, one approach is the use of appetite suppressants. These compounds are frequently amphetamine congeners such as diethylpropion (DEP), phentermine (PHEN), and bupropion (BUP), whose effects are mediated through serotonin, norepinephrine, and dopaminergic pathways. The nucleus accumbens (NAc) shell receives dopaminergic inputs and is involved in feeding and motor activity. However, little is known about how appetite suppressants modulate its activity. Therefore, we characterized behavioral and neuronal NAc shell responses to short-term treatments of DEP, PHEN, and BUP. These compounds caused a transient decrease in weight and food intake while increasing locomotion, stereotypy, and insomnia. They evoked a large inhibitory imbalance in NAc shell spiking activity that correlated with the onset of locomotion and stereotypy. Analysis of the local field potentials (LFPs) showed that all three drugs modulated beta, theta, and delta oscillations. These oscillations do not reflect an aversive-malaise brain state, as ascertained from taste aversion experiments, but tracked both the initial decrease in weight and food intake and the subsequent tolerance to these drugs. Importantly, the appetite suppressant-induced weight loss and locomotion were markedly reduced by intragastric (and intra-NAc shell) infusions of dopamine antagonists SCH-23390 (D1 receptor) or raclopride (D2 receptor). Furthermore, both antagonists attenuated appetite suppressant-induced LFP oscillations and partially restored the imbalance in NAc shell activity. These data reveal that appetite suppressant-induced behavioral and neuronal activity recorded in the NAc shell depend, to various extents, on dopaminergic activation and thus point to an important role for D1/D2-like receptors (in the NAc shell) in the mechanism of action for these anorexic compounds.


Asunto(s)
Depresores del Apetito/farmacología , Antagonistas de los Receptores de Dopamina D2/farmacología , Núcleo Accumbens/efectos de los fármacos , Receptores de Dopamina D1/antagonistas & inhibidores , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Depresores del Apetito/efectos adversos , Benzazepinas/farmacología , Bupropión/efectos adversos , Bupropión/farmacología , Dietilpropión/efectos adversos , Dietilpropión/farmacología , Interacciones Farmacológicas , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Núcleo Accumbens/fisiología , Fentermina/efectos adversos , Fentermina/farmacología , Racloprida/farmacología , Distribución Aleatoria , Ratas Sprague-Dawley , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Trastornos del Inicio y del Mantenimiento del Sueño/inducido químicamente , Conducta Estereotipada/efectos de los fármacos , Conducta Estereotipada/fisiología , Pérdida de Peso/efectos de los fármacos , Pérdida de Peso/fisiología
6.
PLoS One ; 19(4): e0300544, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38656972

RESUMEN

Obesity is a major global health epidemic that has adverse effects on both the people affected as well as the cost to society. Several anti-obesity drugs that target GLP-1 receptors have recently come to the market. Here, we describe the effects of tesofensine, a novel anti-obesity drug that acts as a triple monoamine neurotransmitter reuptake inhibitor. Using various techniques, we investigated its effects on weight loss and underlying neuronal mechanisms in mice and rats. These include behavioral tasks, DeepLabCut videotaped analysis, electrophysiological ensemble recordings, optogenetic activation, and chemogenetic silencing of GABAergic neurons in the Lateral Hypothalamus (LH). We found that tesofensine induces a greater weight loss in obese rats than lean rats, while differentially modulating the neuronal ensembles and population activity in LH. In Vgat-ChR2 and Vgat-IRES-cre transgenic mice, we found for the first time that tesofensine inhibited a subset of LH GABAergic neurons, reducing their ability to promote feeding behavior, and chemogenetically silencing them enhanced tesofensine's food-suppressing effects. Unlike phentermine, a dopaminergic appetite suppressant, tesofensine causes few, if any, head-weaving stereotypy at therapeutic doses. Most importantly, we found that tesofensine prolonged the weight loss induced by 5-HTP, a serotonin precursor, and blocked the body weight rebound that often occurs after weight loss. Behavioral studies on rats with the tastant sucrose indicated that tesofensine's appetite suppressant effects are independent of taste aversion and do not directly affect the perception of sweetness or palatability of sucrose. In summary, our data provide new insights into the effects of tesofensine on weight loss and the underlying neuronal mechanisms, suggesting that tesofensine may be an effective treatment for obesity and that it may be a valuable adjunct to other appetite suppressants to prevent body weight rebound.


Asunto(s)
Fármacos Antiobesidad , Compuestos Bicíclicos Heterocíclicos con Puentes , Neuronas GABAérgicas , Obesidad , Animales , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Ratas , Ratones , Fármacos Antiobesidad/farmacología , Masculino , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Conducta Alimentaria/efectos de los fármacos , Área Hipotalámica Lateral/efectos de los fármacos , Área Hipotalámica Lateral/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Ratones Transgénicos , Pérdida de Peso/efectos de los fármacos , Ratas Sprague-Dawley
7.
Am J Physiol Regul Integr Comp Physiol ; 305(3): R252-70, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23678029

RESUMEN

Despite decades of study, it remains a matter of controversy as to whether in rats taste identification is a rapid process that occurs in about 250-600 ms (one to three licks) or a slow process that evolves over seconds. To address this issue, we trained rats to perform a taste-cued two-response discrimination task (2-RDT). It was found that, after learning, regardless of intensity, the delivery of 10 µl of a tastant (e.g., NaCl or monopotassium glutamate, MPG) was sufficient to identify its taste with maximal accuracy within 400 ms. However, despite overtraining, rats rarely stopped licking in one lick. Thus, a one-drop lick reaction task was developed in which subjects had to rapidly stop licking after release of a stop signal (tastants including water) to obtain rewards. The faster they stopped licking, the greater the reward. Rats did not stop licking after receiving either hedonically positive or negative stop signals, and thus failed to maximize rewards even when reinforced with even larger rewards. In fact, the higher the sucrose concentration given as a stop signal, the greater the number of consummatory licks elicited. However, with a stop signal of 2 mM quinine HCl, they stopped licking in ~370 ms, a time faster than that for sucrose or water, thus showing that in this rapid period, quinine HCl evoked an unpalatable response. Indeed, only when rats licked an empty sipper tube would they usually elicit a single lick to obtain a reward (operant licking). In summary, these data indicate that within 400 ms, taste identification and palatability, must either occur simultaneously or with marked overlap.


Asunto(s)
Conducta Consumatoria/fisiología , Preferencias Alimentarias/fisiología , Aprendizaje/fisiología , Desempeño Psicomotor/fisiología , Recompensa , Gusto/fisiología , Animales , Condicionamiento Operante/fisiología , Señales (Psicología) , Discriminación en Psicología/fisiología , Masculino , Ratas , Ratas Sprague-Dawley
8.
Elife ; 122023 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-37646772

RESUMEN

A new platform for studying how brain activity is linked to behavior enables researchers to perform diverse experiments on mice that have their heads immobilized.


Asunto(s)
Movimiento , Investigación , Animales , Ratones , Humanos , Encéfalo , Investigadores
9.
Cell Rep ; 42(11): 113365, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37924513

RESUMEN

The insular cortex (IC) has been linked to the processing of interoceptive and exteroceptive signals associated with addictive behavior. However, whether the IC modulates the acquisition of drug-related affective states by direct top-down connectivity with ventral tegmental area (VTA) dopamine neurons is unknown. We found that photostimulation of VTA terminals of the anterior insular cortex (aIC) induces rewarding contextual memory, modulates VTA activity, and triggers dopamine release within the VTA. Employing neuronal recordings and neurochemical and transsynaptic tagging techniques, we disclose the functional top-down organization tagging the aIC pre-synaptic neuronal bodies and identifying VTA recipient neurons. Furthermore, systemic administration of amphetamine altered the VTA excitability of neurons modulated by the aIC projection, where photoactivation enhances, whereas photoinhibition impairs, a contextual rewarding behavior. Our study reveals a key circuit involved in developing and retaining drug reward-related contextual memory, providing insight into the neurobiological basis of addictive behavior and helping develop therapeutic addiction strategies.


Asunto(s)
Neuronas Dopaminérgicas , Área Tegmental Ventral , Neuronas Dopaminérgicas/fisiología , Área Tegmental Ventral/fisiología , Corteza Insular , Anfetamina/farmacología , Recompensa
10.
J Neurophysiol ; 108(6): 1739-51, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22745464

RESUMEN

Neurons in the nucleus accumbens (NAc) have been shown to participate in several behavioral states, including feeding and sleep. However, it is not known if the same neuron participates in both states and, if so, how similar are the responses. In addition, since the NAc contains several cell types, it is not known if each type participates in the transitions associated with feeding and sleep. Such knowledge is important for understanding the interaction between two different neural networks. For these reasons we recorded ensembles of NAc neurons while individual rats volitionally transitioned between the following states: awake and goal directed, feeding, quiet-awake, and sleeping. We found that during both feeding and sleep states, the same neurons could increase their activity (be activated) or decrease their activity (be inactivated) by feeding and/or during sleep, thus indicating that the vast majority of NAc neurons integrate sleep and feeding signals arising from spatially distinct neural networks. In contrast, a smaller population was modulated by only one of the states. For the majority of neurons in either state, we found that when one population was excited, the other was inhibited, suggesting that they act as a local circuit. Classification of neurons into putative interneurons [fast-spiking interneurons (pFSI) and choline acetyltransferase interneurons (pChAT)] and projection medium spiny neurons (pMSN) showed that all three types are modulated by transitions to and from feeding and sleep states. These results show, for the first time, that in the NAc, those putative inhibitory interneurons respond similarly to pMSN projection neurons and demonstrate interactions between NAc networks involved in sleep and feeding.


Asunto(s)
Neuronas Colinérgicas/fisiología , Conducta Alimentaria/fisiología , Neuronas GABAérgicas/fisiología , Interneuronas/fisiología , Núcleo Accumbens/fisiología , Sueño/fisiología , Potenciales de Acción , Animales , Mapeo Encefálico , Objetivos , Masculino , Red Nerviosa , Núcleo Accumbens/citología , Ratas , Ratas Sprague-Dawley
11.
Neuroscience ; 493: 81-98, 2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35504509

RESUMEN

The palatability and concentration of sweet foods promote hedonic feeding beyond homeostatic need. Understanding how neurons respond to sweet taste is thus of great importance. The dorsomedial nucleus accumbens shell (dNAcMed) is considered a "sensory sentinel," promoting hedonic feeding. However, it is unknown how neurons in the lateral part (NAcLat) respond to oral sucrose stimulation. Using in vivo calcium imaging of individual D1 and D2 cells in NAcLat of mice performing behavioral licking tasks, we find that D1 and D2 neurons do not act as single homogeneous populations. Instead, their responses are organized into ensembles with context-dependent temporal dynamics around licking sucrose. At the macrostructure of licking (meals), D1 and D2 population activity recorded on the first day predict the licking behavior on subsequent days. However, at the level of the microstructure of licking (bouts), calcium activity increased concurrently in D1 and D2 neurons prior to licking bouts, whereas during licking, calcium signals decreased. Importantly, in a Brief Access Taste Task, calcium responses for D1 and D2 exhibit much more heterogeneity than during a freely licking task. Specifically, D1 and D2 neurons form distinct ensembles: some ramp up in anticipation of the first lick, some respond at the end of the taste-access period, and some categorize sucrose concentrations as low or high. Collectively, NAcLat D1 and D2 neurons are organized in ensembles that adapt to the behavioral context to monitor task-relevant events and sucrose concentrations.


Asunto(s)
Conducta Alimentaria , Núcleo Accumbens , Sacarosa , Animales , Calcio/metabolismo , Conducta Alimentaria/fisiología , Ratones , Neuronas/metabolismo , Núcleo Accumbens/metabolismo , Receptores de Dopamina D1/metabolismo , Sacarosa/administración & dosificación
12.
eNeuro ; 9(3)2022.
Artículo en Inglés | MEDLINE | ID: mdl-35715209

RESUMEN

How do animals experience brain manipulations? Optogenetics has allowed us to manipulate selectively and interrogate neural circuits underlying brain function in health and disease. However, little is known about whether mice can detect and learn from arbitrary optogenetic perturbations from a wide range of brain regions to guide behavior. To address this issue, mice were trained to report optogenetic brain perturbations to obtain rewards and avoid punishments. Here, we found that mice can perceive optogenetic manipulations regardless of the perturbed brain area, rewarding effects, or the stimulation of glutamatergic, GABAergic, and dopaminergic cell types. We named this phenomenon optoception, a perceptible signal internally generated from perturbing the brain, as occurs with interoception. Using optoception, mice can learn to execute two different sets of instructions based on the laser frequency. Importantly, optoception can occur either activating or silencing a single cell type. Moreover, stimulation of two brain regions in a single mouse uncovered that the optoception induced by one brain region does not necessarily transfer to a second not previously stimulated area, suggesting a different sensation is experienced from each site. After learning, they can indistinctly use randomly interleaved perturbations from both brain regions to guide behavior. Collectively taken, our findings revealed that mice's brains could "monitor" perturbations of their self-activity, albeit indirectly, perhaps via interoception or as a discriminative stimulus, opening a new way to introduce information to the brain and control brain-computer interfaces.


Asunto(s)
Encéfalo , Optogenética , Animales , Encéfalo/fisiología , Cabeza , Ratones , Percepción , Recompensa
13.
Front Cell Neurosci ; 16: 823220, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35360496

RESUMEN

Taste memory involves storing information through plasticity changes in the neural network of taste, including the insular cortex (IC) and ventral tegmental area (VTA), a critical provider of dopamine. Although a VTA-IC dopaminergic pathway has been demonstrated, its role to consolidate taste recognition memory remains poorly understood. We found that photostimulation of dopaminergic neurons in the VTA or VTA-IC dopaminergic terminals of TH-Cre mice improves the salience to consolidate a subthreshold novel taste stimulus regardless of its hedonic value, without altering their taste palatability. Importantly, the inhibition of the D1-like receptor into the IC impairs the salience to facilitate consolidation of an aversive taste recognition memory. Finally, our results showed that VTA photostimulation improves the salience to consolidate a conditioned taste aversion memory through the D1-like receptor into the IC. It is concluded that the dopamine activity from the VTA into IC is required to increase the salience enabling the consolidation of a taste recognition memory. Notably, the D1-like receptor activity into the IC is required to consolidate both innate and learned aversive taste memories but not appetitive taste memory.

14.
J Neurosci ; 30(1): 287-303, 2010 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-20053910

RESUMEN

Animals learn which foods to ingest and which to avoid. Despite many studies, the electrophysiological correlates underlying this behavior at the gustatory-reward circuit level remain poorly understood. For this reason, we measured the simultaneous electrical activity of neuronal ensembles in the orbitofrontal cortex, insular cortex, amygdala, and nucleus accumbens while rats licked for taste cues and learned to perform a taste discrimination go/no-go task. This study revealed that rhythmic licking entrains the activity in all these brain regions, suggesting that the animal's licking acts as an "internal clock signal" against which single spikes can be synchronized. That is, as animals learned a go/no-go task, there were increases in the number of licking coherent neurons as well as synchronous spiking between neuron pairs from different brain regions. Moreover, a subpopulation of gustatory cue-selective neurons that fired in synchrony with licking exhibited a greater ability to discriminate among tastants than nonsynchronized neurons. This effect was seen in all four recorded areas and increased markedly after learning, particularly after the cue was delivered and before the animals made a movement to obtain an appetitive or aversive tastant. Overall, these results show that, throughout a large segment of the taste-reward circuit, appetitive and aversive associative learning improves spike-timing precision, suggesting that proficiency in solving a taste discrimination go/no-go task requires licking-induced neural ensemble synchronous activity.


Asunto(s)
Sincronización Cortical , Señales (Psicología) , Aprendizaje Discriminativo/fisiología , Red Nerviosa/fisiología , Recompensa , Gusto/fisiología , Animales , Conducta Animal/fisiología , Condicionamiento Operante/fisiología , Masculino , Ratas , Ratas Long-Evans
15.
IUBMB Life ; 63(10): 824-30, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21901814

RESUMEN

The ability to control neuronal activity using light pulses and optogenetic tools has revealed new properties of neural circuits and established causal relationships between activation of a single genetically defined population of neurons and complex behaviors. Here, we briefly review the causal effect of activity of six genetically defined neural circuits on behavior, including the dopaminergic neurons DA in the ventral tegmental area (VTA); the two main populations of medium-sized spiny neurons (D1- and D2-positive) in the striatum; the giant Cholinergic interneurons in the ventral striatum; and the hypocretin- and MCH- expressing neurons in the lateral hypothalamus. We argue that selective spatiotemporal recruitment and coordinated spiking activity among these cell type-specific neural circuits may underlie the neural integration of reward, learning, arousal and feeding.


Asunto(s)
Nivel de Alerta/fisiología , Ganglios Basales/fisiología , Conducta Alimentaria/fisiología , Área Hipotalámica Lateral/fisiología , Neuronas/fisiología , Recompensa , Área Tegmental Ventral/fisiología , Técnicas Genéticas , Humanos , Vías Nerviosas/fisiología , Óptica y Fotónica , Estimulación Luminosa
16.
Compr Physiol ; 11(4): 2489-2523, 2021 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-34558667

RESUMEN

The gustatory system detects and informs us about the nature of various chemicals we put in our mouth. Some of these have nutritive value (sugars, amino acids, salts, and fats) and are appetitive and avidly ingested, whereas others (atropine, quinine, nicotine) are aversive and rapidly rejected. However, the gustatory system is mainly responsible for evoking the perception of a limited number of qualities that humans taste as sweet, umami, bitter, sour, salty, and perhaps fat [free fatty acids (FFA)] and starch (malto-oligosaccharides). The complex flavors and mouthfeel that we experience while eating food result from the integration of taste, odor, texture, pungency, and temperature. The latter three arise primarily from the somatosensory (trigeminal) system. The sensory organs used for detecting and transducing many chemicals are found in taste buds (TBs) located throughout the tongue, soft palate esophagus, and epiglottis. In parallel with the taste system, the trigeminal nerve innervates the peri-gemmal epithelium to transmit temperature, mechanical stimuli, and painful or cooling sensations such as those produced by changes in temperature as well as from chemicals like capsaicin and menthol, respectively. This article gives an overview of the current knowledge about these TB cells' anatomy and physiology and their trigeminal induced sensations. We then discuss how taste is represented across gustatory cortices using an intermingled and spatially distributed population code. Finally, we review postingestion processing (interoception) and central integration of the tongue-gut-brain interaction, ultimately determining our sensations as well as preferences toward the wholesomeness of nutritious foods. © 2021 American Physiological Society. Compr Physiol 11:1-35, 2021.


Asunto(s)
Papilas Gustativas , Gusto , Encéfalo , Humanos , Quinina , Lengua
17.
Neuron ; 51(4): 483-94, 2006 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-16908413

RESUMEN

The motivation to start or terminate a meal involves the continual updating of information on current body status by central gustatory and reward systems. Previous electrophysiological and neuroimaging investigations revealed region-specific decreases in activity as the subject's state transitions from hunger to satiety. By implanting bundles of microelectrodes in the lateral hypothalamus, orbitofrontal cortex, insular cortex, and amygdala of hungry rats that voluntarily eat to satiety, we have measured the behavior of neuronal populations through the different phases of a complete feeding cycle (hunger-satiety-hunger). Our data show that while most satiety-sensitive units preferentially responded to a unique hunger phase within a cycle, neuronal populations integrated single-unit information in order to reflect the animal's motivational state across the entire cycle, with higher activity levels during the hunger phases. This distributed population code might constitute a neural mechanism underlying meal initiation under different metabolic states.


Asunto(s)
Mapeo Encefálico , Neuronas/clasificación , Neuronas/fisiología , Saciedad/fisiología , Potenciales de Acción/fisiología , Ciclos de Actividad/fisiología , Animales , Conducta Animal , Glucemia , Encéfalo/citología , Conducta Alimentaria/fisiología , Insulina/sangre , Masculino , Ratas , Ratas Long-Evans , Gusto/fisiología , Factores de Tiempo
18.
eNeuro ; 7(5)2020.
Artículo en Inglés | MEDLINE | ID: mdl-33077494

RESUMEN

The intensity of sucrose (its perceived concentration) and its palatability (positive hedonic valence associated with ingestion) are two taste attributes that increase its attractiveness and overconsumption. Although both sensory attributes covary, in that increases in sucrose concentration leads to similar increases in its palatability, this covariation does not imply that they are part of the same process or whether they represent separate processes. Both these possibilities are considered in the literature. For this reason, we tested whether sucrose's perceived intensity could be separated from its hedonically positive palatability. To address this issue, rats were trained in a sucrose intensity task to report the perceived intensity of a range of sucrose concentrations before and after its palatability was changed using a conditioned taste aversion (CTA) protocol. We found that the subjects' performance remained essentially unchanged, although its palatability was changed from hedonically positive to negative. Overall, these data demonstrate that sucrose's perceived intensity and its positive palatability can be dissociated, meaning that changes of one taste attribute render the other mostly unaffected. Thus, the intensity attribute is sufficient to inform the perceptual judgments of sucrose's concentrations.


Asunto(s)
Percepción del Gusto , Gusto , Animales , Condicionamiento Clásico , Preferencias Alimentarias , Ratas , Sacarosa
19.
Front Neurosci ; 14: 572328, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33177980

RESUMEN

D-norpseudoephedrine (NPE), also known as cathine, is found naturally in the shrub Catha edulis "Khat." NPE has been widely used as an appetite suppressant for the treatment of obesity. Although it is known that NPE acts on α1-adrenergic receptors, there is little information about the role of dopamine receptors on NPE's induced anorectic and weight loss effects. Equally untouched is the question of how NPE modulates neuronal activity in the nucleus accumbens shell (NAcSh), a brain reward center, and a pharmacological target for many appetite suppressants. To do this, in rats, we characterized the pharmacological effects induced by NPE on weight loss, food intake, and locomotion. We also determined the involvement of dopamine D1- and D2-like receptors using systemic and intra-NAcSh antagonists, and finally, we recorded single-unit activity in the NAcSh in freely moving rats. We found that NPE decreased 24-h food intake, induced weight loss, and as side effects increased locomotor activity and wakefulness. Also, intraperitoneal and intra-NAcSh administration of D1 and D2 dopamine antagonists partially reversed NPE's induced weight loss and food intake suppression. Furthermore, the D1 antagonist, SCH-23390, eliminated NPE-induced locomotion, whereas the D2 antagonist, raclopride, only delayed its onset. We also found that NPE evoked a net activation imbalance in NAcSh that propelled the population activity trajectories into a dynamic pharmacological brain state, which correlated with the onset of NPE-induced wakefulness. Together, our data demonstrate that NPE modulates NAcSh spiking activity and that both dopamine D1 and D2 receptors are necessary for NPE's induced food intake suppression and weight loss.

20.
Front Neurosci ; 14: 608047, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33551725

RESUMEN

Sucrose is attractive to most species in the animal kingdom, not only because it induces a sweet taste sensation but also for its positive palatability (i.e., oromotor responses elicited by increasing sucrose concentrations). Although palatability is such an important sensory attribute, it is currently unknown which cell types encode and modulate sucrose's palatability. Studies in mice have shown that activation of GABAergic LHAVgat+ neurons evokes voracious eating; however, it is not known whether these neurons would be driving consumption by increasing palatability. Using optrode recordings, we measured sucrose's palatability while VGAT-ChR2 transgenic mice performed a brief access sucrose test. We found that a subpopulation of LHAVgat+ neurons encodes palatability by increasing (or decreasing) their activity as a function of the increment in licking responses evoked by sucrose concentrations. Optogenetic gain of function experiments, where mice were able to choose among available water, 3% and 18% sucrose solutions, uncovered that opto-stimulation of LHAVgat+ neurons consistently promoted higher intake of the most palatable stimulus (18% sucrose). In contrast, if they self-stimulated near the less palatable stimulus, some VGAT-ChR2 mice preferred water over 18% sucrose. Unexpectedly, activation of LHAVgat+ neurons increased quinine intake but only during water deprivation, since in sated animals, they failed to promote quinine intake or tolerate an aversive stimulus. Conversely, these neurons promoted overconsumption of sucrose when it was the nearest stimulus. Also, experiments with solid foods further confirmed that these neurons increased food interaction time with the most palatable food available. We conclude that LHAVgat+ neurons increase the drive to consume, but it is potentiated by the palatability and proximity of the tastant.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA